1
|
Li H, Wu Y, Zhou Q, Wu P, Xing Y, Zhuang Z, Zhao X, Zhang X. Polysaccharide from steamed Polygonatum sibiricum ameliorates ulcerative colitis by protecting the intestinal mucosal barrier and regulating gut microbiota. Int J Biol Macromol 2025; 301:140343. [PMID: 39880230 DOI: 10.1016/j.ijbiomac.2025.140343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/02/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Steamed Polygonatum sibiricum is widely applied in clinical practice for its tonic effect on gastrointestinal tract. A novel polysaccharide named PSSP-EF was extracted from the steamed roots of Polygonatum sibiricum using hot water extraction, ethanol precipitation, and chromatographic purification. PSSP-EF, with a molecular weight of 2.24 × 104 Da, was consisted of mannose, glucosamine hydrochloride, glucose, galactose, xylose, and arabinose in a molar ratio of 10.25: 0.26: 3.56: 80.55: 1.72: 3.66, and its main chain was constituted by→4)-β-D-Galp-(1→4)-β-D-Galp-(1→ and →4)-β-D-Manp-(1→4)-β-D-2ace-Manp-(1→ residues, with branching from β-D-Galp-(1→ residues. PSSP-EF could dramatically relieve clinical symptoms of ulcerative colitis (UC) in mice. Treatment with PSSP-EF significantly alleviated colon inflammation (TNF-α, IL-1β, and IL-6), repaired intestinal mucosal barrier (Occludin, ZO-1, and Claudin-1) and regulated the balance of gut microbiota by increased the levels of Muribaculaeae, while decreasing the levels of Bacteroides, Erysipelatoclostridium, and Romboutsia. Notably, PSSP-EF remarkably increased the levels of acetic, propionic, isobutyric, butyric, valeric, and isovaleric acid in the cecal contents of UC mice. In conclusion, PSSP-EF has a significant therapeutic effect on UC by balancing gut microbiota, protecting intestinal mucosal barrier, and regulating short-chain fatty acid production, and can be developed as a functional food.
Collapse
Affiliation(s)
- Haoran Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yi Wu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qiao Zhou
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Peng Wu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yue Xing
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ziming Zhuang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Xuelan Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
2
|
Gao C, Feng Z, Wang L, Fu K, Yang Z, Wang S, Yu S. A comparative study on in vitro models of necrotizing enterocolitis induced by single and combined stimulation. Arab J Gastroenterol 2025:S1687-1979(24)00097-2. [PMID: 39818482 DOI: 10.1016/j.ajg.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/11/2024] [Accepted: 08/24/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND AND STUDY AIMS Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease in neonates. In vitro model is an indispensable tool to study the pathogenesis of NEC. This study explored the effects of different stress factors on intestinal injury in vitro. MATERIAL AND METHODS Rat intestinal epithelial cell line-6 (IEC-6) cells were exposed to different stressors, including lipopolysaccharide (LPS), cobalt chloride (CoCl2), and a combination of both. We estimated the cell viability, the expression of inflammatory cytokines and tight junction proteins. RESULTS The decrease in IEC-6 cell viability was observed after stimulation by CoCl2 alone or in combination with LPS, but not after stimulation with LPS alone. The expression of interleukin6 (IL-6) and tumornecrosisfactoralpha (TNFα) increased in each group. After stimulation with CoCl2 alone or in combination with LPS, a decrease in Claudin-1 was observed, but an increase was detected after stimulation with LPS alone. Zona occludens 1 (ZO-1) decreased in both mRNA and protein levels after combined stimulation. CONCLUSION The combined stimulation of LPS and CoCl2 on IEC-6 cells could simultaneously induce severe inflammation and barrier damage, which may better simulate the pathological process of NEC.
Collapse
Affiliation(s)
- Chuchu Gao
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, PR China; Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Suzhou, PR China
| | - Zongtai Feng
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Suzhou, PR China
| | - Lixia Wang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Kai Fu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Zuming Yang
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Suzhou, PR China
| | - Sannan Wang
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Suzhou, PR China
| | - Shenglin Yu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, PR China.
| |
Collapse
|
3
|
Mirfeizi Z, Mahmoudi M, Jokar MH, Sahebari M, Noori E, Mehrad-Majd H, Barati M, Faridzadeh A. Impact of synbiotics on disease activity in systemic lupus erythematosus: Results from a randomized clinical trial. J Food Sci 2024; 89:9835-9845. [PMID: 39437223 DOI: 10.1111/1750-3841.17460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects various organs in the body. In SLE, inflammatory cytokines play a crucial role in initiating and sustaining the inflammatory process. Synbiotics may help modulate these inflammatory cytokines. This randomized, double-blind, placebo-controlled clinical trial aimed to assess the impact of synbiotics intervention on interleukin-17A (IL-17A) levels, disease activity, and inflammatory factors in patients with SLE. Fifty SLE patients were randomly assigned to receive either standard therapy plus synbiotics (consisting of Streptococcus thermophilus, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus salivarius, Lactobacillus reuteri, Bifidobacterium lactis, Bifidobacterium longum, Bifidobacterium bifidum, and the prebiotic fructooligosaccharides) or standard therapy alone for 2 months. The results demonstrated a significant reduction in both protein and mRNA levels of IL-17A, as well as in the Systemic Lupus Erythematosus Disease Activity Index 2000 score, within the synbiotics group after the intervention compared to baseline. In contrast, the placebo group did not experience significant changes in IL-17A levels or disease activity. Synbiotic supplementation shows potential as an adjunctive therapeutic approach for SLE management; however, further research is needed to elucidate its underlying mechanisms. PRACTICAL APPLICATION: This study explores the use of synbiotics as a supplementary treatment for systemic lupus erythematosus, which is typically managed with immunosuppressive therapies.
Collapse
Affiliation(s)
- Zahra Mirfeizi
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hassan Jokar
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Sahebari
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Noori
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hasan Mehrad-Majd
- Clinical Research Development Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Barati
- Department of Laboratory Sciences, School of Paramedicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Arezoo Faridzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Xie L, Zhuang Z, Guo B, Huang Y, Shi X, Huang Z, Xu Z, Chen Y, Cao Y, Zheng Y, Wu R, Ma S. Ketamine induced gut microbiota dysbiosis and barrier and hippocampal dysfunction in rats. iScience 2024; 27:111089. [PMID: 39493883 PMCID: PMC11530865 DOI: 10.1016/j.isci.2024.111089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/02/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a pivotal role in drug addiction. However, the pathophysiological mechanism of MGBA in ketamine addiction remains elusive. The present study investigated the ketamine-induced gut microbiota disorders, intestinal barrier dysfunction, and the alterations in brain function, using a conditioned place preference (CPP) model of ketamine addiction in rats. Compared with the control group, ketamine induced decreased amplitude of low-frequency fluctuation (ALFF) values in the hippocampus, and pyknotic nuclei and concentrated cytoplasm in hippocampal neurons, as well as alterations in gut microbiota composition, shortened ileum villi, and thinner colonic mucosa. We also found that the abundance of gut microbiota exhibited correlations with CPP score, hippocampal ALFF value, length of ileum villi, and thickness of colonic mucosa. Our findings provide evidence for abnormal alterations in the MGBA of ketamine-addicted rats, which improves our understating of the mechanism of ketamine addiction and the potential for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lei Xie
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zelin Zhuang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Baowen Guo
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuehua Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Xiaoyan Shi
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Zikai Huang
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Ziquan Xu
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanbin Chen
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yuyin Cao
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yanmin Zheng
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Renhua Wu
- Department of Radiology, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Shuhua Ma
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Laboratory of Molecular Imaging & Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
5
|
Choi H, Kwak MJ, Kang AN, Mun D, Lee S, Park MR, Oh S, Kim Y. Limosilactobacillus fermentum SLAM 216-Derived Extracellular Vesicles Promote Intestinal Maturation in Mouse Organoid Models. J Microbiol Biotechnol 2024; 34:2091-2099. [PMID: 39252638 PMCID: PMC11540603 DOI: 10.4014/jmb.2405.05028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024]
Abstract
Probiotics, when consumed in adequate amounts, can promote the health of the host and beneficially modulate the host's immunity. Particularly during the host's early life, the gut intestine undergoes a period of epithelial maturation in which epithelial cells organize into specific crypt and villus structures. This process can be mediated by the gut microbiota. Recent studies have reported that the administration of probiotics can further promote intestinal maturation in the neonatal intestine. Therefore, in this study, we investigated the effects of extracellular vesicles derived from the Limosilactobacillus fermentum SLAM 216 strain, which is an established probiotic with known immune and anti-aging effects on intestinal epithelial maturation and homeostasis, using mouse small intestinal organoids. As per our findings, treatment with L. fermentum SLAM 216-derived LF216EV (LF216EV) has significantly increased the bud number and size of organoid buds. Furthermore, extracellular vesicle (EV) treatment upregulated the expression of maturation-related genes, including Ascl2, Ephb2, Lgr5, and Sox9. Tight junctions are known to have an important role in the intestinal immune barrier, and EV treatment has significantly increased the expression of genes associated with tight junctions, such as Claudin, Muc2, Occludin, and Zo-1, indicating that it can promote intestinal development. This was supported by RNA sequencing, which revealed the upregulation of genes associated with cAMP-mediated signaling, which is known to regulate cellular processes including cell differentiation. Additionally, organoids exposed to LF216EV exhibited upregulation of genes associated with maintaining brain memory and neurotransmission, suggesting possible future functional implications.
Collapse
Affiliation(s)
- Hyejin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Suengwon Lee
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mi Ri Park
- Korea Food Research Institute, Wanju 55365, Republic of Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju 55069, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
Huang X, Bao J, Yang M, Li Y, Liu Y, Zhai Y. The role of Lactobacillus plantarum in oral health: a review of current studies. J Oral Microbiol 2024; 16:2411815. [PMID: 39444695 PMCID: PMC11497578 DOI: 10.1080/20002297.2024.2411815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/09/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Background Oral non-communicable diseases, particularly dental caries and periodontal disease, impose a significant global health burden. The underlying microbial dysbiosis is a prominent factor, driving interest in strategies that promote a balanced oral microbiome. Lactobacillus plantarum, a gram-positive lactic acid bacterium known for its adaptability, has gained attention for its potential to enhance oral health. Recent studies have explored the use of probiotic L. plantarum in managing dental caries, periodontal disease, and apical periodontitis. However, a comprehensive review on its effects in this context is still lacking. Aims This narrative review evaluates current literature on L. plantarum's role in promoting oral health and highlights areas for future research. Content In general, the utilization of L. plantarum in managing non-communicable biofilm-dependent oral diseases is promising, but additional investigations are warranted. Key areas for future study include: exploring its mechanisms of action, identifying optimal strains or strain combinations of L. plantarum, determining effective delivery methods and dosages, developing commercial antibacterial agents from L. plantarum, and addressing safety considerations related to its use in oral care.
Collapse
Affiliation(s)
- Xinyan Huang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Jianhang Bao
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Mingzhen Yang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Yingying Li
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Youwen Liu
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| |
Collapse
|
7
|
Kazemian N, Pakpour S. Understanding the impact of the gut microbiome on opioid use disorder: Pathways, mechanisms, and treatment insights. Microb Biotechnol 2024; 17:e70030. [PMID: 39388360 PMCID: PMC11466222 DOI: 10.1111/1751-7915.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
The widespread use of opioids for chronic pain management not only poses a significant public health issue but also contributes to the risk of tolerance, dependence, and addiction, leading to opioid use disorder (OUD), which affects millions globally each year. Recent research has highlighted a potential bidirectional relationship between the gut microbiome and OUD. This emerging perspective is critical, especially as the opioid epidemic intensifies, emphasizing the need to investigate how OUD may alter gut microbiome dynamics and vice versa. Understanding these interactions could reveal new insights into the mechanisms of addiction and tolerance, as well as provide novel approaches for managing and potentially mitigating OUD impacts. This comprehensive review explores the intricate bidirectional link through the gut-brain axis, focusing on how opiates influence microbial composition, functional changes, and gut mucosal integrity. By synthesizing current findings, the review aims to inspire new strategies to combat the opioid crisis and leverage microbiome-centred interventions for preventing and treating OUD.
Collapse
Affiliation(s)
- Negin Kazemian
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| | - Sepideh Pakpour
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| |
Collapse
|
8
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
9
|
Abdulqadir R, Al-Sadi R, Haque M, Gupta Y, Rawat M, Ma TY. Bifidobacterium bifidum Strain BB1 Inhibits Tumor Necrosis Factor-α-Induced Increase in Intestinal Epithelial Tight Junction Permeability via Toll-Like Receptor-2/Toll-Like Receptor-6 Receptor Complex-Dependent Stimulation of Peroxisome Proliferator-Activated Receptor γ and Suppression of NF-κB p65. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1664-1683. [PMID: 38885924 PMCID: PMC11372998 DOI: 10.1016/j.ajpath.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Bifidobacterium bifidum strain BB1 causes a strain-specific enhancement in intestinal epithelial tight junction (TJ) barrier. Tumor necrosis factor (TNF)-α induces an increase in intestinal epithelial TJ permeability and promotes intestinal inflammation. The major purpose of this study was to delineate the protective effect of BB1 against the TNF-α-induced increase in intestinal TJ permeability and to unravel the intracellular mechanisms involved. TNF-α produces an increase in intestinal epithelial TJ permeability in Caco-2 monolayers and in mice. Herein, the addition of BB1 inhibited the TNF-α increase in Caco-2 intestinal TJ permeability and mouse intestinal permeability in a strain-specific manner. BB1 inhibited the TNF-α-induced increase in intestinal TJ permeability by interfering with TNF-α-induced enterocyte NF-κB p50/p65 and myosin light chain kinase (MLCK) gene activation. The BB1 protective effect against the TNF-α-induced increase in intestinal permeability was mediated by toll-like receptor-2/toll-like receptor-6 heterodimer complex activation of peroxisome proliferator-activated receptor γ (PPAR-γ) and PPAR-γ pathway inhibition of TNF-α-induced inhibitory kappa B kinase α (IKK-α) activation, which, in turn, resulted in a step-wise inhibition of NF-κB p50/p65, MLCK gene, MLCK kinase activity, and MLCK-induced opening of the TJ barrier. In conclusion, these studies unraveled novel intracellular mechanisms of BB1 protection against the TNF-α-induced increase in intestinal TJ permeability. The current data show that BB1 protects against the TNF-α-induced increase in intestinal epithelial TJ permeability via a PPAR-γ-dependent inhibition of NF-κB p50/p65 and MLCK gene activation.
Collapse
Affiliation(s)
- Raz Abdulqadir
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| | - Rana Al-Sadi
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Mohammad Haque
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Yash Gupta
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Manmeet Rawat
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania
| | - Thomas Y Ma
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Hershey, Pennsylvania.
| |
Collapse
|
10
|
Haque M, Kaminsky L, Abdulqadir R, Engers J, Kovtunov E, Rawat M, Al-Sadi R, Ma TY. Lactobacillus acidophilus inhibits the TNF-α-induced increase in intestinal epithelial tight junction permeability via a TLR-2 and PI3K-dependent inhibition of NF-κB activation. Front Immunol 2024; 15:1348010. [PMID: 39081324 PMCID: PMC11286488 DOI: 10.3389/fimmu.2024.1348010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Background Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Mohammad Haque
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Lauren Kaminsky
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Raz Abdulqadir
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Jessica Engers
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Evgeny Kovtunov
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Manmeet Rawat
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Rana Al-Sadi
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Thomas Y. Ma
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
11
|
Xu Y, Zhang H, Yang H, Liu C, Song C, Cheng Y, He C, Zou Z, Zhou D, Wu G, Zhang X. Eicosapentaenoic acid and docosahexaenoic acid suppress colonic tumorigenesis in obese mice. J Funct Foods 2024; 116:106164. [DOI: 10.1016/j.jff.2024.106164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
12
|
Zhang Y, Anderson RC, You C, Purba A, Yan M, Maclean P, Liu Z, Ulluwishewa D. Lactiplantibacillus plantarum ST-III and Lacticaseibacillus rhamnosus KF7 Enhance the Intestinal Epithelial Barrier in a Dual-Environment In Vitro Co-Culture Model. Microorganisms 2024; 12:873. [PMID: 38792703 PMCID: PMC11124027 DOI: 10.3390/microorganisms12050873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Intestinal barrier hyperpermeability, which is characterised by impaired tight junction proteins, is associated with a variety of gastrointestinal and systemic diseases. Therefore, maintaining intestinal barrier integrity is considered one of the effective strategies to reduce the risk of such disorders. This study aims to investigate the potential benefits of two probiotic strains (Lactiplantibacillus plantarum ST-III and Lacticaseibacillus rhamnosus KF7) on intestinal barrier function by using a physiologically relevant in vitro model of the intestinal epithelium. Our results demonstrate that both strains increased transepithelial electrical resistance, a measure of intestinal barrier integrity. Immunolocalisation studies indicated that this improvement in barrier function was not due to changes in the co-localisation of the tight junction (TJ) proteins ZO-1 and occludin. However, we observed several modifications in TJ-related genes in response to the probiotics, including the upregulation of transmembrane and cytosolic TJ proteins, as well as TJ signalling proteins. Gene expression modulation was strain- and time-dependent, with a greater number of differentially expressed genes and higher fold-change being observed in the L. plantarum ST-III group and at the latter timepoint. Further studies to investigate how the observed gene expression changes can lead to enhanced barrier function will aid in the development of probiotic foods to help improve intestinal barrier function.
Collapse
Affiliation(s)
- Yilin Zhang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, China; (Y.Z.); (C.Y.); (M.Y.)
| | - Rachel C. Anderson
- AgResearch, Te Ohu Rangahau Kai, Palmerston North 4410, New Zealand; (R.C.A.); (A.P.)
| | - Chunping You
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, China; (Y.Z.); (C.Y.); (M.Y.)
| | - Ajitpal Purba
- AgResearch, Te Ohu Rangahau Kai, Palmerston North 4410, New Zealand; (R.C.A.); (A.P.)
| | - Minghui Yan
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, China; (Y.Z.); (C.Y.); (M.Y.)
| | - Paul Maclean
- AgResearch, Grasslands Research Centre, Palmerston North 4410, New Zealand;
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai 200436, China; (Y.Z.); (C.Y.); (M.Y.)
| | - Dulantha Ulluwishewa
- AgResearch, Te Ohu Rangahau Kai, Palmerston North 4410, New Zealand; (R.C.A.); (A.P.)
| |
Collapse
|
13
|
Constantin M, Chifiriuc MC, Mihaescu G, Corcionivoschi N, Burlibasa L, Bleotu C, Tudorache S, Mitache MM, Filip R, Munteanu SG, Gradisteanu Pircalabioru G. Microbiome and cancer: from mechanistic implications in disease progression and treatment to development of novel antitumoral strategies. Front Immunol 2024; 15:1373504. [PMID: 38715617 PMCID: PMC11074409 DOI: 10.3389/fimmu.2024.1373504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/08/2024] [Indexed: 05/23/2024] Open
Abstract
Cancer is a very aggressive disease and one of mankind's most important health problems, causing numerous deaths each year. Its etiology is complex, including genetic, gender-related, infectious diseases, dysbiosis, immunological imbalances, lifestyle, including dietary factors, pollution etc. Cancer patients also become immunosuppressed, frequently as side effects of chemotherapy and radiotherapy, and prone to infections, which further promote the proliferation of tumor cells. In recent decades, the role and importance of the microbiota in cancer has become a hot spot in human biology research, bringing together oncology and human microbiology. In addition to their roles in the etiology of different cancers, microorganisms interact with tumor cells and may be involved in modulating their response to treatment and in the toxicity of anti-tumor therapies. In this review, we present an update on the roles of microbiota in cancer with a focus on interference with anticancer treatments and anticancer potential.
Collapse
Affiliation(s)
- Marian Constantin
- Institute of Biology, Bucharest of Romanian Academy, Bucharest, Romania
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, Bucharest, Romania
- Faculty of Biology, University of Bucharest, Bucharest, Romania
| | | | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast, United Kingdom
- Faculty of Bioengineering of Animal Resources, Banat University of Agricultural Sciences and Veterinary Medicine-King Michael I of Romania, Timisoara, Romania
- Romanian Academy of Scientists, Bucharest, Romania
| | | | - Coralia Bleotu
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, Bucharest, Romania
- Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Sorin Tudorache
- Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | | | - Roxana Filip
- Faculty of Medicine and Biological Sciences, Stefan cel Mare University of Suceava, Suceava, Romania
- Suceava Emergency County Hospital, Suceava, Romania
| | | | - Gratiela Gradisteanu Pircalabioru
- Life, Environmental and Earth Sciences Division, Research Institute of the University of Bucharest, Bucharest, Romania
- Faculty of Biology, University of Bucharest, Bucharest, Romania
- Romanian Academy of Scientists, Bucharest, Romania
- eBio-Hub Research Centre, National University of Science and Technology Politehnica Bucharest, Bucharest, Romania
| |
Collapse
|
14
|
Koh YC, Chang YC, Lin WS, Leung SY, Chen WJ, Wu SH, Wei YS, Gung CL, Chou YC, Pan MH. Efficacy and Mechanism of the Action of Live and Heat-Killed Bacillus coagulans BC198 as Potential Probiotic in Ameliorating Dextran Sulfate Sodium-Induced Colitis in Mice. ACS OMEGA 2024; 9:10253-10266. [PMID: 38463297 PMCID: PMC10918820 DOI: 10.1021/acsomega.3c07529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Inflammatory bowel disease alters the gut microbiota, causes defects in mucosal barrier function, and leads to dysregulation of the immune response to microbial stimulation. This study investigated and compared the efficacy of a candidate probiotic strain, Bacillus coagulans BC198, and its heat-killed form in treating dextran sulfate sodium-induced colitis. Both live and heat-killed B. coagulans BC198 increased gut barrier-associated protein expression, reduced neutrophil and M1 macrophage infiltration of colon tissue, and corrected gut microbial dysbiosis induced by colitis. However, only live B. coagulans BC198 could alleviate the general symptoms of colitis, prevent colon shortening, and suppress inflammation and tissue damage. At the molecular level, live B. coagulans BC198 was able to inhibit Th17 cells while promoting Treg cells in mice with colitis, reduce pro-inflammatory MCP-1 production, and increase anti-inflammatory IL-10 expression in the colonic mucosa. The live form of B. coagulans BC198 functioned more effectively than the heat-killed form in ameliorating colitis by enhancing the anti-inflammatory response and promoting Treg cell accumulation in the colon.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Ya-Chu Chang
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Sheng Lin
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Food Science, National Quemoy University, Quemoy 892, Taiwan
| | - Siu-Yi Leung
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Jen Chen
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Shiuan-Huei Wu
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Yu-Shan Wei
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Chiau-Ling Gung
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Ya-Chun Chou
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung City 40402, Taiwan
- Department
of Health and Nutrition Biotechnology, Asia
University, Taichung City 41354, Taiwan
| |
Collapse
|
15
|
So YJ, Park OJ, Kwon Y, Im J, Lee D, Yun SH, Cho K, Yun CH, Han SH. Bacillus subtilis Induces Human Beta Defensin-2 Through its Lipoproteins in Human Intestinal Epithelial Cells. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10224-4. [PMID: 38376819 DOI: 10.1007/s12602-024-10224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
Human intestinal epithelial cells (IECs) play an important role in maintaining gut homeostasis by producing antimicrobial peptides (AMPs). Bacillus subtilis, a commensal bacterium, is considered a probiotic. Although its protective effects on intestinal health are widely reported, the key component of B. subtilis responsible for its beneficial effects remains elusive. In this study, we tried to identify the key molecules responsible for B. subtilis-induced AMPs and their molecular mechanisms in a human IEC line, Caco-2. B. subtilis increased human beta defensin (HBD)-2 mRNA expression in a dose- and time-dependent manner. Among the B. subtilis microbe-associated molecular patterns, lipoprotein (LPP) substantially increased the mRNA expression and protein production of HBD-2, whereas lipoteichoic acid and peptidoglycan did not show such effects. Those results were confirmed in primary human IECs. In addition, both LPP recognition and HBD-2 secretion mainly took place on the apical side of fully differentiated and polarized Caco-2 cells through Toll-like receptor 2-mediated JNK/p38 MAP kinase/AP-1 and NF-κB pathways. HBD-2 efficiently inhibited the growth of the intestinal pathogens Staphylococcus aureus and Bacillus cereus. Furthermore, LPPs pre-incubated with lipase or proteinase K decreased LPP-induced HBD-2 expression, suggesting that the lipid and protein moieties of LPP are crucial for HBD-2 expression. Q Exactive Plus mass spectrometry identified 35 B. subtilis LPP candidates within the LPP preparation, and most of them were ABC transporters. Taken together, these results suggest that B. subtilis promotes HBD-2 secretion in human IECs mainly with its LPPs, which might enhance the protection from intestinal pathogens.
Collapse
Affiliation(s)
- Yoon Ju So
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeongkag Kwon
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung-Ho Yun
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Kun Cho
- Center for Research Equipment, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
- Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Judkins TC, Solch-Ottaiano RJ, Ceretto-Clark B, Nieves C, Colee J, Wang Y, Tompkins TA, Caballero-Calero SE, Langkamp-Henken B. The effect of an acute aspirin challenge on intestinal permeability in healthy adults with and without prophylactic probiotic consumption: a double-blind, placebo-controlled, randomized trial. BMC Gastroenterol 2024; 24:4. [PMID: 38166769 PMCID: PMC10759586 DOI: 10.1186/s12876-023-03102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Healthy individuals may experience increases in intestinal permeability after chronic or acute use of non-steroidal anti-inflammatory drugs, which may be attenuated by probiotics. This study investigates the effects of an acute aspirin challenge on gastroduodenal barrier function with or without prophylactic probiotic consumption. METHODS Twenty-nine generally healthy participants (26 ± 6 years) completed a 14-week randomized, double-blind, crossover trial. A probiotic containing 2 Lactobacilli strains or placebo was administered for 3 weeks, with a 4-week washout period between crossover phases. Daily and weekly questionnaires assessing gastrointestinal function were completed for 2 weeks before until 2 weeks after each intervention to assess gastrointestinal function. Gastroduodenal permeability was assessed by urinary excretion of orally administered sucrose after 1, 2, and 3 weeks of each intervention with a 1950 mg-aspirin challenge after 2 weeks of supplementation. Stool samples were collected weekly during supplementation for detection of species of interest. RESULTS Gastroduodenal permeability increased with aspirin challenge (Week 1: 3.4 ± 0.6 μmol vs Week 2: 9.9 ± 1.0 μmol urinary sucrose; p < 0.05). There were no differences in the change in permeability after the aspirin challenge or gastrointestinal function between interventions. CONCLUSION The acute aspirin challenge significantly increased intestinal permeability similarly in both groups, and prophylactic probiotic consumption was unable to prevent the loss in this particular model.
Collapse
Affiliation(s)
- Taylor C Judkins
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA
| | - Rebecca J Solch-Ottaiano
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA
| | - Brendan Ceretto-Clark
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA
| | - Carmelo Nieves
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA
| | - James Colee
- IFAS Statistical Consulting Unit, University of Florida, PO Box 110500, Gainesville, FL, 32611-0500, USA
| | - Yu Wang
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA
- Citrus Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Lake Alfred, FL, 33850, USA
| | - Thomas A Tompkins
- Lallemand Bio-Ingredients, 1620 Rue Prefontaine, Montreal, QC, H1N 2W8, Canada
| | | | - Bobbi Langkamp-Henken
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr, Gainesville, FL, 32611, USA.
| |
Collapse
|
17
|
Conn KA, Borsom EM, Cope EK. Implications of microbe-derived ɣ-aminobutyric acid (GABA) in gut and brain barrier integrity and GABAergic signaling in Alzheimer's disease. Gut Microbes 2024; 16:2371950. [PMID: 39008552 PMCID: PMC11253888 DOI: 10.1080/19490976.2024.2371950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024] Open
Abstract
The gut microbial ecosystem communicates bidirectionally with the brain in what is known as the gut-microbiome-brain axis. Bidirectional signaling occurs through several pathways including signaling via the vagus nerve, circulation of microbial metabolites, and immune activation. Alterations in the gut microbiota are implicated in Alzheimer's disease (AD), a progressive neurodegenerative disease. Perturbations in gut microbial communities may affect pathways within the gut-microbiome-brain axis through altered production of microbial metabolites including ɣ-aminobutyric acid (GABA), the primary inhibitory mammalian neurotransmitter. GABA has been shown to act on gut integrity through modulation of gut mucins and tight junction proteins and may be involved in vagus nerve signal inhibition. The GABAergic signaling pathway has been shown to be dysregulated in AD, and may be responsive to interventions. Gut microbial production of GABA is of recent interest in neurological disorders, including AD. Bacteroides and Lactic Acid Bacteria (LAB), including Lactobacillus, are predominant producers of GABA. This review highlights how temporal alterations in gut microbial communities associated with AD may affect the GABAergic signaling pathway, intestinal barrier integrity, and AD-associated inflammation.
Collapse
Affiliation(s)
- Kathryn A. Conn
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Emily M. Borsom
- Center for Data-Driven Discovery for Biology, Allen Institute, Seattle, WA, USA
| | - Emily K. Cope
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| |
Collapse
|
18
|
Zhou Y, Duan L, Zeng Y, Song X, Pan K, Niu L, Pu Y, Li J, Khalique A, Fang J, Jing B, Zeng D, Shen B, Ni X. The panda-derived Lactiplantibacillus plantarum BSG201683 improves LPS-induced intestinal inflammation and epithelial barrier disruption in vitro. BMC Microbiol 2023; 23:249. [PMID: 37674107 PMCID: PMC10481503 DOI: 10.1186/s12866-023-02928-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/03/2023] [Indexed: 09/08/2023] Open
Abstract
Captive pandas are suffering from intestinal infection due to intestinal microbiota characterized by a high abundance of Enterobacteriaceae induced by long-term captivity. Probiotic supplements showed improvement in intestinal barrier function and inflammation. However, the effects of panda-derived probiotics on the intestinal epithelium and inflammation have not been elucidated. In the present study, lipopolysaccharide (LPS) impaired Caco-2 and RAW264.7 inflammatory models were applied to assess the protection of Lactiplantibacillus plantarum BSG201683 (L. plantarum G83) on barrier disruption and inflammation. The results showed that treatment with L. plantarum G83 significantly decreased the paracellular permeability to fluorescein isothiocyanate conjugated dextran (MW 4000, FITC-D4) after LPS induction. Meanwhile, L. plantarum G83 alleviated the reduction in tight junction (TJ) proteins and downregulated proinflammatory cytokines caused by LPS in Caco-2 cells. L. plantarum G83 also significantly decreased the expression and secretion of pro-inflammatory cytokines in LPS-induced RAW264.7 cells. In addition, the IL-10 increased in both Caco-2 and RAW264.7 cells after L. plantarum G83 treatment. The phagocytosis activity of RAW264.7 cells was significantly increased after L. plantarum G83 treatment. Toll-like receptor 4/ nuclear factor kappa-B (TLR4/NF-κB) signaling pathways were significantly down-regulated after L. plantarum G83 intervention, and the phosphorylation of NF-κB/p65 was consistent with this result. Our findings suggest that L. plantarum G83 improves intestinal inflammation and epithelial barrier disruption in vitro.
Collapse
Affiliation(s)
- Yi Zhou
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China
| | - Ling Duan
- Animal Feed Affairs of Sichuan Province, Sichuan Provincial Department of Agriculture and Rural Affairs, Chengdu, 610041, Sichuan, China
| | - Yan Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xu Song
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Lili Niu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, 610081, Sichuan, China
| | - Yang Pu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, 610081, Sichuan, China
| | - Jiakun Li
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jing Fang
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bo Jing
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Dong Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Bairong Shen
- Department of Urology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 611130, Sichuan, China.
| | - Xueqin Ni
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
19
|
Yan C, Qu H, Li X, Feng B. Holothurian Wall Hydrolysate Ameliorates Cyclophosphamide-Induced Immunocompromised Mice via Regulating Immune Response and Improving Gut Microbiota. Int J Mol Sci 2023; 24:12583. [PMID: 37628768 PMCID: PMC10454611 DOI: 10.3390/ijms241612583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Some biologically active compounds isolated from sea cucumbers stimulate the body's immune response by activating immune cells. Immune function is closely related to the integrity intestinal barrier and balanced gut microbiota. However, it is unknown whether the daily administration of holothurian wall hydrolysate (HWH) ameliorated intestinal dysbiosis and barrier injury induced by immunodeficiency. This study aimed to investigate the immunomodulatory effect and the underlying mechanism of HWH in cyclophosphamide (CTX)-induced immunocompromised mice. BALB/c mice received CTX (80 mg/kg, intraperitoneally) once a day for 3 days to induce immunodeficiency, and then they received the oral administration of HWH (80 or 240 mg/kg) or levamisole hydrochloride (LH, 40 mg/kg, positive control), respectively, once a day for 7 days. We utilized 16S rRNA sequencing for microbial composition alterations, histopathological analysis for splenic and colonic morphology, Western blotting for expressions of tight junction proteins (TJs), and quantitative real-time (qRT)-PCR for measurements of pro-inflammatory cytokines. HWH attenuated the immune organ damage induced by CTX, increased the secretions of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α, and promoted the recovery of goblet cells and the production of TJs (claudin-1, occludin, and ZO-1) in the colon of the immunocompromised mice. Moreover, HWH promoted the growth of beneficial microorganisms such as Lactobacillus, Lachnospiraceae, Christensenellaceae, and Bifidobacterium, while it suppressed the populations of Ruminococcus, Staphylococcus, and Streptococcus. These results demonstrate that HWH elicits intestinal mucosal immunity, repairs the damage to intestinal mucosal integrity, and normalizes the imbalanced intestinal microbial profiles in immunocompromised mice. It may be helpful to identify the biological activities of HWH to support its potential use in new prebiotics, immunomodulatory agents, and medical additives for intestinal repair.
Collapse
Affiliation(s)
| | | | - Xinli Li
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (C.Y.); (H.Q.)
| | - Bin Feng
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China; (C.Y.); (H.Q.)
| |
Collapse
|
20
|
Yin Y, Liao Y, Li J, Pei Z, Wang L, Shi Y, Peng H, Tan Y, Li C, Bai H, Ma C, Gong Y, Wei T, Peng H. Lactobacillus plantarum GX17 benefits growth performance and improves functions of intestinal barrier/intestinal flora among yellow-feathered broilers. Front Immunol 2023; 14:1195382. [PMID: 37465686 PMCID: PMC10351386 DOI: 10.3389/fimmu.2023.1195382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Lactobacillus plantarum has recently been found to be a natural source feed additive bacteria with great advantages in food safety and animal welfare. Discovering novel strains with commercial application potentiation could benefit the local poultry industry, and in particular support Chinese farmers. In this study, we tested a recently isolated novel strain of Lactobacillus plantarum GX17 as a feed additive on the growth performance and intestinal barrier functions of 1-day-old Chinese yellow-feather chicks. As good as other commercial probiotics, feeding with Lactobacillus plantarum GX17 showed significant improvements in humoral immune responses and enhanced the immune effect after vaccination for either the Newcastle disease vaccine or the avian influenza vaccine. This study also found that feeding with Lactobacillus plantarum GX17 improved the feed-to-weight ratio and caused a significant increase of the villus length to crypt depth ratio. Furthermore, Lactobacillus plantarum GX17 significantly up-regulated the mRNA expression of CLDN, MUC2, and TLR2, all of which are jejunum-associated barrier genes, indicating an improvement of the intestinal barrier functions by enhancing the tight junction between epithelia cells. These results are comparable to the effects of feeding the commercial complex probiotics that improve the expression levels of CLDN, ocludin, MUC2, TLR2, and TLR4. In terms of maintaining intestinal health, commercial complex probiotics increased the relative abundance of Parabacteroides and Romboutsia, while Lactobacillus plantarum GX17 increased the relative abundance of Pseudoflavonifractor. Our data suggest that Lactobacillus plantarum GX17 could enhance the intestinal absorption of nutrients and therefore improve the growth performance of Chinese yellow-feather chicks. In conclusion, compared with the commercial complex probiotics, Lactobacillus plantarum GX17 has more positive effects on the growth performance and intestinal barrier function of yellow-feather chickens, and can be used as a feed additive.
Collapse
Affiliation(s)
- Yangyan Yin
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yuying Liao
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Jun Li
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Zhe Pei
- Virginia Tech, Department of Engineering, Blacksburg, New York, NY, United States
| | - Leping Wang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yan Shi
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hongyan Peng
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yizhou Tan
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Changting Li
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Huili Bai
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Chunxia Ma
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Yu Gong
- Guizhou Provincial Livestock and Poultry Genetic Resources Management Station, Guiyang, China
| | - Tianchao Wei
- Institute of Animal Science and Technology, Guangxi University, Nanning, China
| | - Hao Peng
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Key Laboratory of China(Guangxi)-Association of Southeast Asian Nations (ASEAN) Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| |
Collapse
|
21
|
Cuna A, Nsumu M, Menden HL, Chavez-Bueno S, Sampath V. The Detrimental Effects of Peripartum Antibiotics on Gut Proliferation and Formula Feeding Injury in Neonatal Mice Are Alleviated with Lactobacillus rhamnosus GG. Microorganisms 2023; 11:1482. [PMID: 37374984 DOI: 10.3390/microorganisms11061482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Peripartum antibiotics can negatively impact the developing gut microbiome and are associated with necrotizing enterocolitis (NEC). The mechanisms by which peripartum antibiotics increase the risk of NEC and strategies that can help mitigate this risk remain poorly understood. In this study, we determined mechanisms by which peripartum antibiotics increase neonatal gut injury and evaluated whether probiotics protect against gut injury potentiated by peripartum antibiotics. To accomplish this objective, we administered broad-spectrum antibiotics or sterile water to pregnant C57BL6 mice and induced neonatal gut injury to their pups with formula feeding. We found that pups exposed to antibiotics had reduced villus height, crypt depth, and intestinal olfactomedin 4 and proliferating cell nuclear antigen compared to the controls, indicating that peripartum antibiotics impaired intestinal proliferation. When formula feeding was used to induce NEC-like injury, more severe intestinal injury and apoptosis were observed in the pups exposed to antibiotics compared to the controls. Supplementation with the probiotic Lactobacillus rhamnosus GG (LGG) reduced the severity of formula-induced gut injury potentiated by antibiotics. Increased intestinal proliferating cell nuclear antigen and activation of the Gpr81-Wnt pathway were noted in the pups supplemented with LGG, suggesting partial restoration of intestinal proliferation by probiotics. We conclude that peripartum antibiotics potentiate neonatal gut injury by inhibiting intestinal proliferation. LGG supplementation decreases gut injury by activating the Gpr81-Wnt pathway and restoring intestinal proliferation impaired by peripartum antibiotics. Our results suggest that postnatal probiotics may be effective in mitigating the increased risk of NEC associated with peripartum antibiotic exposure in preterm infants.
Collapse
Affiliation(s)
- Alain Cuna
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Marianne Nsumu
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Heather L Menden
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Susana Chavez-Bueno
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
- Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Venkatesh Sampath
- Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO 64108, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| |
Collapse
|
22
|
Snyder KB, Hunter CJ. Bugs and the barrier: A review of the gut microbiome and intestinal barrier in necrotizing enterocolitis. Semin Pediatr Surg 2023; 32:151310. [PMID: 37290337 DOI: 10.1016/j.sempedsurg.2023.151310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease that affects premature neonates. It frequently results in significant morbidity and mortality for those affected. Years of research into the pathophysiology of NEC have revealed it to be a variable and multifactorial disease. However, there are risk factors associated with NEC including low birth weight, prematurity, intestinal immaturity, alterations in microbial colonization, and history of rapid or formula based enteral feeds (Fig. 1).1-3 An accepted generalization of the pathogenesis of NEC includes a hyperresponsive immune reaction to insults such as ischemia, starting formula feeds, or alterations in the microbiome with pathologic bacterial colonization and translocation. This reaction causes a hyperinflammatory response disrupting the normal intestinal barrier, allowing abnormal bacterial translocation and ultimately sepsis.1,2,4 This review will focus specifically on the interactions with the microbiome and intestinal barrier function in NEC.
Collapse
Affiliation(s)
- K Brooke Snyder
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States
| | - Catherine J Hunter
- Division of Pediatric Surgery, Oklahoma Children's Hospital, 1200 Everett Drive, ET NP 2320 Oklahoma City, OK 73104, United States; The University of Oklahoma Health Sciences Center, Department of Surgery, 800 Research Parkway, Suite 449, Oklahoma City, OK 73104, United States.
| |
Collapse
|
23
|
Lopez-Escalera S, Lund ML, Hermes GDA, Choi BSY, Sakamoto K, Wellejus A. In Vitro Screening for Probiotic Properties of Lactobacillus and Bifidobacterium Strains in Assays Relevant for Non-Alcoholic Fatty Liver Disease Prevention. Nutrients 2023; 15:nu15102361. [PMID: 37242245 DOI: 10.3390/nu15102361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial metabolic disorder that poses health challenges worldwide and is expected to continue to rise dramatically. NAFLD is associated with metabolic syndrome, type 2 diabetes mellitus, and impaired gut health. Increased gut permeability, caused by disturbance of tight junction proteins, allows passage of damaging microbial components that, upon reaching the liver, have been proposed to trigger the release of inflammatory cytokines and generate cellular stress. A growing body of research has suggested the utilization of targeted probiotic supplements as a preventive therapy to improve gut barrier function and tight junctions. Furthermore, specific microbial interactions and metabolites induce the secretion of hormones such as GLP-1, resulting in beneficial effects on liver health. To increase the likelihood of finding beneficial probiotic strains, we set up a novel screening platform consisting of multiple in vitro and ex vivo assays for the screening of 42 bacterial strains. Analysis of transepithelial electrical resistance response via co-incubation of the 42 bacterial strains with human colonic cells (Caco-2) revealed improved barrier integrity. Then, strain-individual metabolome profiling was performed revealing species-specific clusters. GLP-1 secretion assay with intestinal secretin tumor cell line (STC-1) found at least seven of the strains tested capable of enhancing GLP-1 secretion in vitro. Gene expression profiling in human biopsy-derived intestinal organoids was performed using next generation sequencing transcriptomics post bacterial co-incubation. Here, different degrees of immunomodulation by the increase in certain cytokine and chemokine transcripts were found. Treatment of mouse primary hepatocytes with selected highly produced bacterial metabolites revealed that indole metabolites robustly inhibited de novo lipogenesis. Collectively, through our comprehensive bacterial screening pipeline, not previously ascribed strains from both Lactobacillus and Bifidobacterium genera were proposed as potential probiotics based on their ability to increase epithelial barrier integrity and immunity, promote GLP-1 secretion, and produce metabolites relevant to liver health.
Collapse
Affiliation(s)
- Silvia Lopez-Escalera
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
- Fakultät für Biowissenschaften, Friedrich-Schiller Universität Jena, Bachstraβe 18K, 07743 Jena, Germany
| | - Mari L Lund
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Gerben D A Hermes
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Béatrice S-Y Choi
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anja Wellejus
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| |
Collapse
|
24
|
Golubkova A, Hunter CJ. Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC. Microorganisms 2023; 11:1247. [PMID: 37317221 PMCID: PMC10221463 DOI: 10.3390/microorganisms11051247] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host internal environment resulting in changes in the intestinal barrier. Pathogenic microbial proliferation and breach of the immature intestinal barrier are proposed to be crucial steps in the development of neonatal diseases such as necrotizing enterocolitis. This article will review the current literature on the intestinal barrier in the neonatal gut, the consequences of microbiome development for this defense system, and how prematurity can influence neonatal susceptibility to gastrointestinal infection.
Collapse
Affiliation(s)
| | - Catherine J. Hunter
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
25
|
Birmann PT, Casaril AM, Pesarico AP, Rodrigues RR, Conceição FR, Sousa FSS, Collares T, Seixas FK, Savegnago L. Komagataella pastoris KM71H Mitigates Depressive-Like Phenotype, Preserving Intestinal Barrier Integrity and Modulating the Gut Microbiota in Mice. Mol Neurobiol 2023; 60:4017-4029. [PMID: 37016046 DOI: 10.1007/s12035-023-03326-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/20/2023] [Indexed: 04/06/2023]
Abstract
The role of intestinal microbiota in the genesis of mental health has received considerable attention in recent years, given that probiotics are considered promising therapeutic agents against major depressive disorder. Komagataella pastoris KM71H is a yeast with probiotic properties and antidepressant-like effects in animal models of depression. Hence, we evaluated the antidepressant-like effects of K. pastoris KM71H in a model of antibiotic-induced intestinal dysbiosis in male Swiss mice. The mice received clindamycin (200 μg, intraperitoneal) and, after 24 h, were treated with K. pastoris KM71H at a dose of 8 log CFU/animal by intragastric administration (ig) or PBS (vehicle, ig) for 14 consecutive days. Afterward, the animals were subjected to behavioral tests and biochemical analyses. Our results showed that K. pastoris KM71H administration decreased the immobility time in the tail suspension test and increased grooming activity duration in the splash test in antibiotic-treated mice, thereby characterizing its antidepressant-like effect. We observed that these effects of K. pastoris KM71H were accompanied by the modulation of the intestinal microbiota, preservation of intestinal barrier integrity, and restoration of the mRNA levels of occludin, zonula occludens-1, zonula occludens-2, and toll-like receptor-4 in the small intestine, and interleukin-1β in the hippocampi of mice. Our findings provide solid evidence to support the development of K. pastoris KM71H as a new probiotic with antidepressant-like effects.
Collapse
Affiliation(s)
- Paloma T Birmann
- Neurobiotechnology Research Group, Graduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Angela M Casaril
- Neurobiotechnology Research Group, Graduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Ana Paula Pesarico
- Neurobiotechnology Research Group, Graduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Rafael R Rodrigues
- Applied Immunology Laboratory, Graduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabricio R Conceição
- Applied Immunology Laboratory, Graduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fernanda Severo Sabedra Sousa
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Graduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Tiago Collares
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Graduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabiana K Seixas
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Graduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Lucielli Savegnago
- Neurobiotechnology Research Group, Graduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, (UFPel), Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
26
|
Rocha SDC, Lei P, Morales-Lange B, Mydland LT, Øverland M. From a cell model to a fish trial: Immunomodulatory effects of heat-killed Lactiplantibacillus plantarum as a functional ingredient in aquafeeds for salmonids. Front Immunol 2023; 14:1125702. [PMID: 36993984 PMCID: PMC10040762 DOI: 10.3389/fimmu.2023.1125702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023] Open
Abstract
Paraprobiotics (dead/inactivated probiotics) are promising candidates in functional feeds to promote growth performance, modulate intestinal microbiota and enhance immune response of fish. During industrial production, fish are exposed to several stressful conditions such as handling, sub-optimal nutrition and diseases that can lead to reduced growth, increased mortalities and large economical losses. Such problems can be mitigated by use of functional feeds, leading to more-sustainable aquaculture and improved animal welfare. Lactiplantibacillus plantarum strain L-137 is a common bacterium found in fermented Southeast Asian dish made from fish and rice. The benefits of its heat-killed form (HK L-137) related to growth performance and immunomodulation have been studied in farmed fish such as Nile Tilapia (Oreochromis niloticus), striped catfish (Pangasianodon hypophthalmus) and bighead catfish (Clarias macrocephalus). To study if such benefits can also be observed in salmonids, we worked both at in vitro level using an intestinal epithelium cell line from rainbow trout (Oncorhynchus mykiss; RTgutGC) stimulated with HK L-137 (Feed LP20™) and at in vivo level with pre-smolt Atlantic salmon (Salmo salar) fed HK L-137 at different inclusion levels (20, 100 and 500 mg of Feed LP20™ kg-1 feed). In RTgutGC, the results showed that the barrier function of the cell monolayer was strengthened along with an increased production of IL-1β and a decreased production of Anxa1, indicating a modulation of the immune response. Interestingly, a similar trend was detected at the in vivo level in distal intestine from fish fed the highest inclusion level of HK L-137. Here, a lower production of Anxa1 was also detected (after a 61-day feeding period) in addition to an increase of total plasma IgM in the same group. Furthermore, the RNA-seq analysis showed that HK L-137 was able to modulate the gene expression of pathways related to molecular function, biological process and cellular component in distal intestine, without compromising fish performance and gut microbiota. Taken together, our study has shown that HK L-137 can modulate physiological response of Atlantic salmon, making fish more robust against stressful conditions during production.
Collapse
|
27
|
Meijer S, Pasquinelli E, Renzi S, Lavasani S, Nouri M, Erlandsson L, Cavalieri D, Hansson SR. Gut Micro- and Mycobiota in Preeclampsia: Bacterial Composition Differences Suggest Role in Pathophysiology. Biomolecules 2023; 13:346. [PMID: 36830715 PMCID: PMC9953204 DOI: 10.3390/biom13020346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Preeclampsia is a severe pregnancy-related inflammatory disease without an effective treatment. The pathophysiology remains partly unknown. However, an increased inflammatory response and oxidative stress are part of the maternal systemic reaction. Recent data have suggested that dysbiosis of the gut microbiome plays a role in preeclampsia as well as other inflammatory diseases. However, dysbiosis in preeclampsia has not been studied in a Scandinavian population. Furthermore, although the fungal flora may also have anti-inflammatory properties, it has never been studied in preeclampsia. We included 25 preeclamptic and 29 healthy third-trimester women for the ITS and 16S sequencing of fungal and bacterial microbiota, respectively. Calprotectin was measured to assess systemic and intestinal inflammatory responses. The fungal diversity differed with BMI and gestational length, suggesting a link between fungi and the immune changes seen in pregnancy. An LEfSe analysis showed 18 significantly differentially abundant bacterial taxa in PE, including enriched Bacteroidetes and depleted Verrucomicrobia and Syntergistota at the phylum level and depleted Akkermansia at the genus level, suggesting a role in the pathophysiology of PE.
Collapse
Affiliation(s)
- Sofie Meijer
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, 22242 Lund, Sweden
| | - Elena Pasquinelli
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Sonia Renzi
- Department of Biology, University of Florence, 50121 Florence, Italy
| | - Shahram Lavasani
- Department of Biology, Lund University, 22362 Lund, Sweden
- ImmuneBiotech AB, Medicon Village, 22381 Lund, Sweden
| | - Mehrnaz Nouri
- ImmuneBiotech AB, Medicon Village, 22381 Lund, Sweden
| | - Lena Erlandsson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden
| | - Duccio Cavalieri
- Department of Biology, University of Florence, 50121 Florence, Italy
| | - Stefan R. Hansson
- Division of Obstetrics and Gynecology, Department of Clinical Sciences Lund, Lund University, 22185 Lund, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, 22242 Lund, Sweden
| |
Collapse
|
28
|
Gao X, Yu J, Chang L, Wang Y, Sun X, Mu G, Qian F. In vitro antibacterial activity of Bacillus coagulans T242 on Caco-2 cells infected with Salmonella Typhimurium. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
29
|
Aleman RS, Moncada M, Aryana KJ. Leaky Gut and the Ingredients That Help Treat It: A Review. Molecules 2023; 28:619. [PMID: 36677677 PMCID: PMC9862683 DOI: 10.3390/molecules28020619] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/31/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023] Open
Abstract
The human body is in daily contact with potentially toxic and infectious substances in the gastrointestinal tract (GIT). The GIT has the most significant load of antigens. The GIT can protect the intestinal integrity by allowing the passage of beneficial agents and blocking the path of harmful substances. Under normal conditions, a healthy intestinal barrier prevents toxic elements from entering the blood stream. However, factors such as stress, an unhealthy diet, excessive alcohol, antibiotics, and drug consumption can compromise the composition of the intestinal microbiota and the homeostasis of the intestinal barrier function of the intestine, leading to increased intestinal permeability. Intestinal hyperpermeability can allow the entry of harmful agents through the junctions of the intestinal epithelium, which pass into the bloodstream and affect various organs and systems. Thus, leaky gut syndrome and intestinal barrier dysfunction are associated with intestinal diseases, such as inflammatory bowel disease and irritable bowel syndrome, as well as extra-intestinal diseases, including heart diseases, obesity, type 1 diabetes mellitus, and celiac disease. Given the relationship between intestinal permeability and numerous conditions, it is convenient to seek an excellent strategy to avoid or reduce the increase in intestinal permeability. The impact of dietary nutrients on barrier function can be crucial for designing new strategies for patients with the pathogenesis of leaky gut-related diseases associated with epithelial barrier dysfunctions. In this review article, the role of functional ingredients is suggested as mediators of leaky gut-related disorders.
Collapse
Affiliation(s)
- Ricardo Santos Aleman
- School of Nutrition and Food Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA 28081, USA
| | - Marvin Moncada
- Department of Food, Bioprocessing & Nutrition Sciences and the Plants for Human Health Institute, North Carolina State University, North Carolina Research Campus, Kannapolis, NC 27599, USA
| | - Kayanush J. Aryana
- School of Nutrition and Food Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA 28081, USA
| |
Collapse
|
30
|
Prebiotic activity of chitooligosaccharides and their ability to alleviate necrotizing enterocolitis in newborn rats. Carbohydr Polym 2023; 299:120156. [PMID: 36876780 DOI: 10.1016/j.carbpol.2022.120156] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022]
Abstract
Chitooligosaccharides (COS) have many bioactive functions and favorable prospects in the fields of biomedicine and functional foods. In this study, COS was found to significantly improve the survival rate of neonatal necrotizing enterocolitis (NEC) model rats, alter the composition of the intestinal microbiota, inhibit the expression of inflammatory cytokines, and alleviate intestinal pathological injury. In addition, COS also increased the abundance of Akkermansia, Bacteroides, and Clostridium sensu stricto 1 in the intestines of normal rats (the normal rat model is more universal). The in vitro fermentation results found that COS was degraded by the human gut microbiota to promote the abundance of Clostridium sensu stricto 1 and produced numerous short-chain fatty acids (SCFAs). In vitro metabolomic analysis revealed that COS catabolism was associated with significant increases in 3-hydroxybutyrate acid and γ-aminobutyric acid. This study provides evidence for the potential of COS as a prebiotic in food products and to ameliorate NEC development in neonatal rats.
Collapse
|
31
|
Américo MF, Freitas ADS, da Silva TF, de Jesus LCL, Barroso FAL, Campos GM, Santos RCV, Gomes GC, Assis R, Ferreira Ê, Mancha-Agresti P, Laguna JG, Chatel JM, Carvalho RDDO, Azevedo V. Growth differentiation factor 11 delivered by dairy Lactococcus lactis strains modulates inflammation and prevents mucosal damage in a mice model of intestinal mucositis. Front Microbiol 2023; 14:1157544. [PMID: 37138633 PMCID: PMC10149842 DOI: 10.3389/fmicb.2023.1157544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Mucositis is an inflammation of the gastrointestinal mucosa that debilitate the quality of life of patients undergoing chemotherapy treatments. In this context, antineoplastic drugs, such as 5-fluorouracil, provokes ulcerations in the intestinal mucosa that lead to the secretion of pro-inflammatory cytokines by activating the NF-κB pathway. Alternative approaches to treat the disease using probiotic strains show promising results, and thereafter, treatments that target the site of inflammation could be further explored. Recently, studies reported that the protein GDF11 has an anti-inflammatory role in several diseases, including in vitro and in vivo results in different experimental models. Hence, this study evaluated the anti-inflammatory effect of GDF11 delivered by Lactococcus lactis strains NCDO2118 and MG1363 in a murine model of intestinal mucositis induced by 5-FU. Our results showed that mice treated with the recombinant lactococci strains presented improved histopathological scores of intestinal damage and a reduction of goblet cell degeneration in the mucosa. It was also observed a significant reduction of neutrophil infiltration in the tissue in comparison to positive control group. Moreover, we observed immunomodulation of inflammatory markers Nfkb1, Nlrp3, Tnf, and upregulation of Il10 in mRNA expression levels in groups treated with recombinant strains that help to partially explain the ameliorative effect in the mucosa. Therefore, the results found in this study suggest that the use of recombinant L. lactis (pExu:gdf11) could offer a potential gene therapy for intestinal mucositis induced by 5-FU.
Collapse
Affiliation(s)
- Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Tales Fernando da Silva
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- INRAE, Institut Agro Rennes-Angers, STLO, Rennes, France
| | - Luís Cláudio Lima de Jesus
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rhayane Cristina Viegas Santos
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriel Camargos Gomes
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Rafael Assis
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ênio Ferreira
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jean-Marc Chatel
- INRAE, AgroParisTech, MICALIS, Université Paris-Saclay, Jouy-en-Josas, France
| | - Rodrigo Dias de Oliveira Carvalho
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Vasco Azevedo
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Vasco Azevedo,
| |
Collapse
|
32
|
Bai X, Xu Q, Zhang W, Wang C. The Gut-Eye Axis: Correlation Between the Gut Microbiota and Autoimmune Dry Eye in Individuals With Sjögren Syndrome. Eye Contact Lens 2023; 49:1-7. [PMID: 36544282 DOI: 10.1097/icl.0000000000000953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2022] [Indexed: 12/24/2022]
Abstract
ABSTRACT The impact of gut microbiota on human health, autoimmunity, and disease occurrence has long been recognized since the advancement of metagenomic sequencing technology has enabled a new level of perspective on the human microbiome. Emerging findings also suggest the existence of a gut-eye axis, wherein gut dysbiosis may be a crucial factor affecting the onset and progression of multiple ocular diseases. Sjögren syndrome (SS) is a chronic autoimmune disease mainly affecting the exocrine glands, primarily the lacrimal gland in the eye, resulting in severe dry eye. Although there are currently various treatments for environmental dry eye, the efficacy for SS-related autoimmune dry eye is limited, and new and more effective therapies still need to be explored. The latest studies have demonstrated that the gut microbiota plays a key role in the pathogenesis of autoimmune dry eye. This review describes the effect of gut microbiota on the ocular surface of autoimmune dry eye; introduces the presumable pathways forming the "gut dysbiosis-ocular surface-lacrimal gland axis"; discusses the advantages of restoring intestinal microecology to treat dry eye by fecal microbiota transplantation or probiotics, which are expected to provide perspectives into the correlation between the gut microbiome and dry eye; enhance our understanding of the pathogenesis in autoimmune dry eye; and be useful in the development of future interventions of dry eye by regulating the gut microbiota.
Collapse
Affiliation(s)
- Xudong Bai
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | | | | | |
Collapse
|
33
|
Miao Z, Miao Z, Liu M, Xu S. Melatonin ameliorates imidacloprid-induced intestinal injury by negatively regulating the PGN/P38MAPK pathway in the common carp (Cyprinuscarpio). FISH & SHELLFISH IMMUNOLOGY 2022; 131:1063-1074. [PMID: 36375784 DOI: 10.1016/j.fsi.2022.11.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 06/16/2023]
Abstract
Imidacloprid (IMI), one of the most frequently used neonicotinoid insecticides in agriculture, is resided in surface water worldwide and poses a threat to aquatic organisms. Melatonin (MT) provides effective protection against insecticide-induced toxicity, nevertheless, the toxic effects and whether MT attenuates intestinal injury caused by IMI exposure in the common carps remains poorly explored. Previous studies have reported adverse effects of IMI exposure on intestinal health status. Therefore, we first demonstrated that IMI altered the composition and function of the intestinal microbiota, destroying the integrity of intestinal ultrastructure, increasing intestinal permeability. Meanwhile, metagenomic sequencing and ELISA kits results hypothesized that peptidoglycan (PGN) is an IMI-triggered intestinal microbial metabolite. Subsequently, we thus further elucidated that IMI induced an increase in intestinal tight junction permeability by inducing PGN secretion in vitro model. MT addition dramatically attenuated IMI-induced intestinal toxicity by remitting PGN synthesis and thus resecuring tight junction permeability, thereby reducing intestinal injury. SB203580 was supplied as a P38MAPK inhibitor to alleviate the increased permeability of tight junctions induced by IMI/PGN. Therefore, these findings confirmed that MT protects against IMI-induced intestinal injury by negatively regulating PGN/P38MAPK pathway to antagonize the increased tight junction permeability.
Collapse
Affiliation(s)
- Zhiruo Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhiying Miao
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Min Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
34
|
Wang Z, Li L, Wang S, Wei J, Qu L, Pan L, Xu K. The role of the gut microbiota and probiotics associated with microbial metabolisms in cancer prevention and therapy. Front Pharmacol 2022; 13:1025860. [PMID: 36452234 PMCID: PMC9702994 DOI: 10.3389/fphar.2022.1025860] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2023] Open
Abstract
Cancer is the second leading cause of elevated mortality worldwide. Thus, the development of drugs and treatments is needed to enhance the survival rate of the cancer-affected population. Recently, gut microbiota research in the healthy development of the human body has garnered widespread attention. Many reports indicate that changes in the gut microbiota are strongly associated with chronic inflammation-related diseases, including colitis, liver disease, and cancer within the intestine and the extraintestinal tract. Different gut bacteria are vital in the occurrence and development of tumors within the gut and extraintestinal tract. The human gut microbiome has significant implications for human physiology, including metabolism, nutrient absorption, and immune function. Moreover, diet and lifestyle habits are involved in the evolution of the human microbiome throughout the lifetime of the host and are involved in drug metabolism. Probiotics are a functional food with a protective role in cancer development in animal models. Probiotics alter the gut microbiota in the host; thus, beneficial bacterial activity is stimulated, and detrimental activity is inhibited. Clinical applications have revealed that some probiotic strains could reduce the occurrence of postoperative inflammation among cancer patients. An association network was constructed by analyzing the previous literature to explore the role of probiotics from the anti-tumor perspective. Therefore, it provides direction and insights for research on tumor treatment.
Collapse
Affiliation(s)
- Zijun Wang
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Lanqing Li
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Shunshun Wang
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Wei
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Linghang Qu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Lianhong Pan
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Kang Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
35
|
Urlacher SS, Kim EY, Luan T, Young LJ, Adjetey B. Minimally invasive biomarkers in human and non-human primate evolutionary biology: Tools for understanding variation and adaptation. Am J Hum Biol 2022; 34:e23811. [PMID: 36205445 PMCID: PMC9787651 DOI: 10.1002/ajhb.23811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/21/2022] [Accepted: 09/10/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The use of minimally invasive biomarkers (MIBs - physiological biomarkers obtained from minimally invasive sample types) has expanded rapidly in science and medicine over the past several decades. The MIB approach is a methodological strength in the field of human and non-human primate evolutionary biology (HEB). Among humans and our closest relatives, MIBs provide unique opportunities to document phenotypic variation and to operationalize evolutionary hypotheses. AIMS This paper overviews the use of MIBs in HEB. Our objectives are to (1) highlight key research topics which successfully implement MIBs, (2) identify promising yet under-investigated areas of MIB application, and (3) discuss current challenges in MIB research, with suggestions for advancing the field. DISCUSSION AND CONCLUSIONS A range of MIBs are used to investigate focal topics in HEB, including energetics and life history variation/evolution, developmental plasticity, and social status and dominance relationships. Nonetheless, we identify gaps in existing MIB research on traits such as physical growth and gut function that are central to the field. Several challenges remain for HEB research using MIBs, including the need for additional biomarkers and methods of assessment, robust validations, and approaches that are standardized across labs and research groups. Importantly, researchers must provide better support for adaptation and fitness effects in hypothesis testing (e.g., by obtaining complementary measures of energy expenditure, demonstrating redundancy of function, and performing lifetime/longitudinal analyses). We point to continued progress in the use of MIBs in HEB to better understand the past, present, and future of humans and our closest primate relatives.
Collapse
Affiliation(s)
- Samuel S. Urlacher
- Department of AnthropologyBaylor UniversityWacoTexasUSA
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
- Child and Brain Development ProgramCIFARTorontoOntarioCanada
| | - Elizabeth Y. Kim
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
- Department of BiologyBaylor UniversityWacoTexasUSA
| | - Tiffany Luan
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| | - Lauren J. Young
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| | - Brian Adjetey
- Human Evolutionary Biology and Health LabBaylor UniversityWacoTexasUSA
| |
Collapse
|
36
|
Li Y, Zhu J, Lin G, Gao K, Yu Y, Chen S, Chen L, Chen Z, Li L. Probiotic effects of Lacticaseibacillus rhamnosus 1155 and Limosilactobacillus fermentum 2644 on hyperuricemic rats. Front Nutr 2022; 9:993951. [PMID: 36245501 PMCID: PMC9562091 DOI: 10.3389/fnut.2022.993951] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Hyperuricemia is the main cause of gout and involved in the occurrence of multiple diseases, such as hypertension, metabolic disorders and chronic kidney disease. Emerging evidence suggests that lactic acid bacteria (LAB) have shown the beneficial effects on the prevention or treatment of hyperuricemia. In this study, the urate-lowering effect of two LAB strains, Lacticaseibacillus rhamnosus 1155 (LR1155) and Limosilactobacillus fermentum 2644 (LF2644) on hyperuricemic rats were investigated. A hyperuricemic rat model was induced by the intragastric treatment of potassium oxonate, combined with a high purine diet. The oral administration of LR1155, LF2644, or a combination of LR1155 and LF2644 for 4 weeks significantly prevented the rise of the serum uric acid (UA) induced by hyperuricemia. LR1155 and LF2644 significantly elevated the fecal UA levels, increased the UA content and up-regulated gene expression of UA transporter, ATP-binding cassette subfamily G-2 (ABCG2), in colon and jejunum tissues, suggesting the accelerated UA excretion from the intestine. Besides, LR1155 significantly inhibited the activity of xanthine oxidase (XOD) in liver and serum, benefited the reduce of UA production. In addition, LF2644 strengthened the gut barrier functions through an up-regulation of the gene expressions for occluding and mucin2, accompanied with the reduced inflammatory indicators of lipopolysaccharide (LPS) and interleukin-1β (IL-1β) in hyperuricemic rat. Moreover, using 16s rDNA high-throughput sequencing of feces, LR1155 was shown to improve the hyperuricemia induced gut microbial dysbiosis. The genera Roseburia, Butyricicoccus, Prevotella, Oscillibacter, and Bifidobacterium may associate with the effect of LR1155 on microbiota in hyperuricemic rats. Collectively, the results indicated that LR1155 and LF2644 exhibit urate-lowering effects and could be used alone or in combination as a new adjuvant treatment for hyperuricemia.
Collapse
Affiliation(s)
- Yanjun Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Jun Zhu
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- *Correspondence: Jun Zhu,
| | - Guodong Lin
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Kan Gao
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Yunxia Yu
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Su Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Lie Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Zuoguo Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Li Li
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| |
Collapse
|
37
|
Liu Z, Jiang Z, Zhang Z, Liu T, Fan Y, Liu T, Peng N. Bacillus coagulans in Combination with Chitooligosaccharides Regulates Gut Microbiota and Ameliorates the DSS-Induced Colitis in Mice. Microbiol Spectr 2022; 10:e0064122. [PMID: 35900082 PMCID: PMC9430726 DOI: 10.1128/spectrum.00641-22] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Ulcerative colitis (UC) are chronic inflammatory disorders, which may be caused by intestinal barrier dysfunction, immune system disorders and intestinal microbiota dysbiosis. Synbiotic, the combination of probiotics and prebiotics, is thought to be a pragmatic approach in mitigating inflammation in UC. Bacillus coagulans has been recognized as a potential probiotic for treating intestinal diseases because of its favorable industrial and probiotic properties, including sporulation and lactic acid production. In this study, we evaluated the treatment effects of the B. coagulans FCYS01 spores with or without the chitooligosaccharides (COSs) on UC generated using dextran sulfate sodium (DSS) in mice. Supplementation of B. coagulans spores, prebiotic COSs or the synbiotic (the spores + COSs) had a significant positive effect on DSS-induced UC. The disease activity index and histological damage score were significantly reduced after these supplementations. Compared to DSS group, these supplementations also significantly modulated the cytokines IL-4, IL-6, IL-8, IL-10, and C-reactive protein (CRP) levels and significantly maintained expressions of tight junction proteins and mucin protein and promotes recovery of the intestinal barrier. In addition, these supplementations regulate the composition of gut microbiota and improve the production of short-chain fatty acids (SCFAs), through enrichment of SCFA-producing bacteria, such as Akkermansia and Ruminococcus species. In summary, the synbiotic ameliorated the overall inflammatory status of the experimental UC model and showed a better treatment effect than B. coagulans or COSs did alone as revealed by the markers such as, colon length, IL-4 and Occludin levels. IMPORTANCE Probiotic and prebiotic are believed to be useful in alleviating the inflammatory, thereby resolving or preventing the severity of UC. Spore-forming bacteria Bacillus coagulans show advantages of stability and probiotic effects, being suggested as the important probiotics for UC treatment. Here, we demonstrate that administration of B. coagulans spores, chitooligosaccharides (COSs), or the synbiotic attenuates DSS-induced colitis and significantly correlates with altered gut immune responses. The treatment effect of the synbiotic is inferred to be relied on the enrichment of probiotic bacteria, such as Akkermansia and Ruminococcaceae species, which are reported to be crucial important for gut health. Our findings facilitate the development of therapeutic and preventive strategies for UC using spore-forming lactic acid bacteria in combination with COSs.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, People’s Republic of China
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Ziyang Jiang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Zhenting Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Tong Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Yurong Fan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Tao Liu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Nan Peng
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, People’s Republic of China
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| |
Collapse
|
38
|
Kim J, Ahn SW, Kim JY, Whon TW, Lim SK, Ryu BH, Han NS, Choi HJ, Roh SW, Lee SH. Probiotic Lactobacilli ameliorate alcohol-induced hepatic damage via gut microbial alteration. Front Microbiol 2022; 13:869250. [PMID: 36081800 PMCID: PMC9446534 DOI: 10.3389/fmicb.2022.869250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Alcoholic liver disease (ALD), which includes fatty liver, cirrhosis, steatosis, fibrosis, and hepatocellular carcinoma, is a global health problem. The probiotic effects of lactic acid bacteria (LAB) are well-known; however, their protective effect against ALD remains unclear. Therefore, in this study, our objective was to assess the protective effects of LAB on ALD. To this end, mice were fed either a normal diet or an alcohol diet for 10 days (to induce ALD) accompanied by vehicle treatment (the NC and AC groups) or kimchi-derived LAB (Lactiplantibacillus plantarum DSR J266 and Levilactobacillus brevis DSR J301, the AL group; or Lacticaseibacillus rhamnosus GG, the AG group). Our results showed that mice in the AC group showed significantly higher serum aspartate aminotransferase and alanine aminotransferase levels than those in the normal diet groups; however, their levels in the AL and AG groups were relatively lower. We also observed that the AL and AG groups showed relatively lower interleukin-6 levels than the AC group. Additionally, AC group showed the accumulation of several fat vesicles in the liver, while the AL and AG groups showed remarkably lower numbers of fat vesicles. The relative abundance of Enterococcus feacalis, which showed association with liver injury, significantly increased in the AC group compared with its levels in the normal diet groups. However, the AG group showed a decreased relative abundance in this regard, confirming that LAB exerted an improvement effect on gut microbial community. These findings suggested that via gut microbiota alteration, the ingestion of LAB can alleviate the ill effects of alcohol consumption, including inflammation, liver damage, gut dysbiosis, and abnormal intestinal nutrient metabolism.
Collapse
Affiliation(s)
- Juseok Kim
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
- Microbiome Research Team, LISCure Biosciences Inc., Seongnam, South Korea
| | - Seong Woo Ahn
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
- Microbiome Research Team, LISCure Biosciences Inc., Seongnam, South Korea
| | - Joon Yong Kim
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
- Microbiome Research Team, LISCure Biosciences Inc., Seongnam, South Korea
| | - Tae Woong Whon
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
| | - Seul Ki Lim
- Fermentation Regulation Technology Research Group, World Institute of Kimchi, Gwangju, South Korea
| | - Byung Hee Ryu
- Food Research Division, Food BU, Daesang Corporation Research Institute, Icheon, South Korea
| | - Nam Soo Han
- Department of Food Science and Biotechnology, Brain Korea 21 Center for Bio-Health Industry, Chungbuk National University, Cheongju, South Korea
| | - Hak-Jong Choi
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
| | - Seong Woon Roh
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
- Microbiome Research Team, LISCure Biosciences Inc., Seongnam, South Korea
| | - Se Hee Lee
- Kimchi Functionality Research Group, World Institute of Kimchi, Gwangju, South Korea
- *Correspondence: Se Hee Lee,
| |
Collapse
|
39
|
Modulatory Impacts of Multi-Strain Probiotics on Rabbits’ Growth, Nutrient Transporters, Tight Junctions and Immune System to Fight against Listeria monocytogenes Infection. Animals (Basel) 2022; 12:ani12162082. [PMID: 36009671 PMCID: PMC9405287 DOI: 10.3390/ani12162082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Weaning is a crucial period associated with great stress and susceptibility to infection, implying adverse impacts on farmed rabbits’ production. Recently, probiotics have been provided as direct microbial feed supplements, which are considered the ideal antibiotic substitutes during pathogenic infections with an emphasis on promoting rabbits’ growth and modulating their immune functions. Therefore, our experiment was carried out to explore the efficacy of multi-strain probiotics (MSP) on rabbits’ growth, molecular aspects, such as nutrients transporters, cytokines, and intestinal integrity, and effectiveness against Listeria monocytogenes (L. monocytogenes) infection. Altogether, our findings proposed the beneficial consequences of MSP on rabbits’ growth, gut health, and immunity. After post-experimental infection of rabbits with L. monocytogenes, administration of MSP during the whole rearing period greatly reduced the detrimental impact of infection and consequently renovated efficient rabbits’ production. Abstract Multi-strain probiotics (MSP) are considered innovative antibiotics’ substitutes supporting superior gut health and immunity of farmed rabbits. The promising roles of MSP on performance, intestinal immunity, integrity and transporters, and resistance against Listeria monocytogenes (L. monocytogenes) were evaluated. In the feeding trial, 220 rabbits were fed a control diet or diet supplemented with three MSP graded levels. At 60 days of age, rabbits were experimentally infected with L. monocytogenes and the positive control, enrofloxacin, prophylactic MSP (MSPP), and prophylactic and therapeutic MSP (MSPTT) groups were included. During the growing period, MSP at the level of 1 × 108 CFU/kg diet (MSPIII) promoted the rabbits’ growth, upregulated the nutrient transporters and tight-junction-related genes, and modified cytokines expression. Supplementing MSPTT for L. monocytogenes experimentally-infected rabbits restored the impaired growth and intestinal barriers, reduced clinical signs of severity and mortalities, and attenuated the excessive inflammatory reactions. Notably, enrofloxacin decreased L. monocytogenes and beneficial microbial loads; unlike MSPTT, which decreased pathogenic bacterial loads and sustained the beneficial ones. Histopathological changes were greatly reduced in MSPTT, confirming its promising role in restricting L. monocytogenes translocation to different organs. Therefore, our results suggest the use of MSPTT as an alternative to antibiotics, thereby conferring protection for rabbits against L. monocytogenes infection.
Collapse
|
40
|
Miri ST, Sotoodehnejadnematalahi F, Amiri MM, Pourshafie MR, Rohani M. The impact of Lactobacillus and Bifidobacterium probiotic cocktail on modulation of gene expression of gap junctions dysregulated by intestinal pathogens. Arch Microbiol 2022; 204:417. [PMID: 35737111 DOI: 10.1007/s00203-022-03026-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022]
Abstract
Probiotics are special bacterial strains with strain specific impacts. They can affect health condition in intestine by producing organic acid, competing with pathogens and maintaining cells homeostasis. Regarding to importance of cell junctions in cells transportation and the influence of pathogens in their functions which lead to inflammation, the impact of probiotic strains comprised of Lactobacillus and Bifidobacterium strains on two important members of gap junctions (Cx26 and Cx43) were assayed. The expressions of cell junction genes in contact with probiotic cocktail along with pathogenic components of enterotoxigenic Escherichia coli and Salmonella typhimurium on HT-29 cell line in different treatment orders were evaluated. Results analysis demonstrated downregulation of cx26 and cx43 along with pathogenic components while, probiotic cocktail could modulate their expression by upregulation. We concluded that Lactobacillus and Bifidobacterium strains were efficient probiotics, when they were used as one cocktail, impacted grater amount on the expression of cell junctions and this might lead to modulate homeostasis and reveal inflammation symptoms in intestine.
Collapse
Affiliation(s)
- Seyedeh Tina Miri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Mohammad Mehdi Amiri
- Department of Immunology, School of Public Health, Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Reza Pourshafie
- Department of Bacteriology, Pasteur Institute of Iran, No. 358, 12th Farvardin Ave, Jomhhoori St, Tehran, Iran.
| | - Mahdi Rohani
- Department of Bacteriology, Pasteur Institute of Iran, No. 358, 12th Farvardin Ave, Jomhhoori St, Tehran, Iran.
| |
Collapse
|
41
|
Wei CX, Wu JH, Huang YH, Wang XZ, Li JY. Lactobacillus plantarum improves LPS-induced Caco2 cell line intestinal barrier damage via cyclic AMP-PKA signaling. PLoS One 2022; 17:e0267831. [PMID: 35639684 PMCID: PMC9154120 DOI: 10.1371/journal.pone.0267831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 04/17/2022] [Indexed: 11/18/2022] Open
Abstract
Lactobacillus plantarum (LP) has been shown to exhibit protective effects on intestinal barrier function in septic rats, although the regulatory mechanism has not been established. We determined whether LP imparts such protective effects in a lipopolysaccharide (LPS)-induced Caco2 cell monolayer model and whether cAMP-PKA signaling is the underlying mechanism of action. The cyclic adenosine monophosphate (cAMP) agonist, forskolin (FSK), and the protein kinase A (PKA) inhibitor, HT89, were used to study the protective effect of LP on the destruction of the tight junction (TJ) structure of cells treated with LPS and the corresponding changes in cAMP-PKA signaling. Our experimental results demonstrated that LP promoted the expression of TJ proteins between Caco2 cells after LPS treatment, and increased the electrical barrier detection (TEER) between Caco2 cells. Moreover, transmission electron microscopy (TEM) revealed that the TJ structural integrity of cells treated with LPS + LP was improved compared to cells treated with LPS alone. In addition, our findings were consistent between the FSK and LP intervention group, while HT89 inhibited LP influence. Taken together, our results indicate that LP has an improved protective effect on LPS-induced damage to the monolayer membrane barrier function of Caco2 cells and is regulated by the cAMP-PKA pathway.
Collapse
Affiliation(s)
- Chen-Xiang Wei
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, P.R. China
| | - Ju-Hua Wu
- Digestive Endoscopy Center, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, P.R. China
| | - Yue-Hong Huang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, P.R. China
| | - Xiao-Zhong Wang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, P.R. China
- * E-mail: (XZW); (JYL)
| | - Jian-Ying Li
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, P.R. China
- * E-mail: (XZW); (JYL)
| |
Collapse
|
42
|
Bu Y, Liu Y, Liu Y, Wang S, Liu Q, Hao H, Yi H. Screening and Probiotic Potential Evaluation of Bacteriocin-Producing Lactiplantibacillus plantarum In Vitro. Foods 2022; 11:foods11111575. [PMID: 35681325 PMCID: PMC9180163 DOI: 10.3390/foods11111575] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Probiotics are gaining attention due to their functions of regulating the intestinal barrier and promoting human health. The production of bacteriocins is one of the important factors for probiotics to exert beneficial properties. This study aimed to screen bacteriocin-producing Lactiplantibacillus plantarum and evaluate the probiotic potential in vitro. It was found that L. plantarum Q7, L. plantarum F3-2 and L. plantarum YRL45 could produce bacteriocins and inhibit common intestinal pathogens. These three strains had probiotic potential with tolerance to the gastrointestinal environmental and colonization in the gut, and exhibited various degrees of anti-inflammatory activity and tight junction function in the intestinal barrier. Particularly, L. plantarum YRL45 could significantly (p < 0.05) reduce the increase in nitric oxide (NO), prostaglandin E2 (PGE2), necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) induced by lipopolysaccharide (LPS), thereby easing inflammatory response. L. plantarum F3-2 could remarkably (p < 0.05) up-regulate the expression levels of ZO-1, Occludin and Claudin-1 in intestinal epithelial injured cells, which was conducive to protecting the intestinal barrier. These findings provided fundamental information about the probiotic properties of bacteriocin-producing L. plantarum, which suggested that L. plantarum Q7, L. plantarum F3-2 and L. plantarum YRL45 had the potential to be used as novel probiotic strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huaxi Yi
- Correspondence: ; Tel.: +86-0532-13792497030
| |
Collapse
|
43
|
Zhang Y, Wang O, Mi H, Yi J, Cai S. Rhus chinensis Mill. fruits prevent necrotizing enterocolitis in rat pups via regulating the expressions of key proteins involved in multiple signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115103. [PMID: 35157955 DOI: 10.1016/j.jep.2022.115103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Based on ancient records and previous studies, many parts of Rhus chinensis Mill., including the fruits, have good preventive and therapeutic effects on inflammation, malaria, diarrhea, and gastrointestinal diseases. Rhus plants and Galla chinensis produced from R. chinensis leaves can also prevent or cure intestinal diseases. However, the preventive effect and molecular mechanisms of R. chinensis fruits on necrotizing enterocolitis (NEC) have not been comprehensively studied. AIM OF THE STUDY This article aims to estimate the effect of the 80% ethanol extract of R. chinensis fruits (RM) on alleviating NEC in rat pups and illustrate the potential molecular mechanisms. MATERIALS AND METHODS Rat pups were subjected to formula feeding, intermittent hypoxic, and cold stresses to establish the NEC model. The preventive effects of RM on NEC were evaluated through survival rate; clinical sickness index; macroscopic conditions; histopathology; and expression levels of inflammatory markers (i.e., tumor necrosis factor-α [TNF-α], interleukin-6 [IL-6]), oxidative stress indicators (i.e., total antioxidant status [TAS], total oxidant status [TOS], superoxide dismutase [SOD], glutathione peroxidase [GSH-Px], myeloperoxidase [MPO], malondialdehyde [MDA]), and tight junction proteins (i.e., Zonula Occludens 1 [ZO-1], Occludin). Moreover, the expression levels of several key proteins involved in oxidative stress (i.e., nuclear factor erythroid 2-related factor 2 [Nrf2], NAD(P)H-quinone oxidoreductase-1 [NQO1]), inflammation (i.e., Toll-like receptor 4 [TLR4], phosphorylated-nuclear factor kappa-B [p-NF-κB], inducible nitric oxide synthase [iNOS]), and apoptosis (i.e., cleaved cysteinyl aspartate specific proteinase-3 [cleaved Caspase-3], Bcl-2-associated X [Bax], B-cell lymphoma-2 [Bcl-2]) in intestinal tissues were analyzed to clarify the molecular mechanisms. RESULTS The extract particularly high doses (400 mg RM/kg body weight) could remarkably reduce the mortality and clinical sickness score and improve the macroscopic condition and histopathological injury of the intestine in NEC pups. After RM administration, the levels of TOS, TNF-α, IL-6, MPO, and MDA in the bowel tissue decreased, whereas the levels of TAS, SOD, and GSH-Px were significantly enhanced. The expression levels of ZO-1 and Occludin proteins were dramatically augmented in RM-treated groups to maintain intestinal barrier integrity. Further analyses revealed that RM might prevent NEC pups by improving some pivotal proteins involved in oxidative stress, inflammation, and apoptosis of enterocytes, namely, by down-regulating the levels of TLR4, p-NF-κB, iNOS, cleaved Caspase-3, and Bax and up-regulating the levels of Bcl-2, NQO1, and Nrf2. CONCLUSIONS The RM prevented the intestinal inflammation and damage caused by NEC by regulating the expression of several pivotal proteins involved in oxidative stress, inflammation, and apoptosis. This study might provide a scientific basis for R. chinensis fruits as a traditional herbal medicine to prevent and/or alleviate NEC.
Collapse
Affiliation(s)
- Yi Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Ou Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, People's Republic of China
| | - Hongying Mi
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, Yunnan Province, People's Republic of China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, People's Republic of China.
| |
Collapse
|
44
|
Kim DW, Jeong HS, Kim E, Lee H, Choi CH, Lee SJ. Oral delivery of stem-cell-loaded hydrogel microcapsules restores gut inflammation and microbiota. J Control Release 2022; 347:508-520. [PMID: 35597403 DOI: 10.1016/j.jconrel.2022.05.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 05/06/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
Mesenchymal stem cells (MSCs) are an attractive candidate for the treatment of inflammatory bowel disease (IBD), but their poor delivery rate to an inflamed colon is a major factor hampering the clinical potential of stem cell therapies. Moreover, there remains a formidable hurdle to overcome with regard to survival and homing in to injured sites. Here, we develop a strategy utilizing monodisperse hydrogel microcapsules with a thin intermediate oil layer prepared by a triple-emulsion drop-based microfluidic approach as an in-situ oral delivering carrier. The oral delivery of stem-cell-loaded hydrogel microcapsules (SC-HM) enhances MSC survival and retention in the hostile stomach environment due to the intermediate oil layer and low value of the overall stiffness, facilitating programmable cell release during gastrointestinal peristalsis. SC-HM is shown to induce tissue repair, reduce the colonic macrophage infiltration responsible for the secretion of the pro-inflammatory factors, and significantly mitigate the severity of IBD in a mouse model, where MSCs released by SC-HM successfully accumulate at the colonic crypt. Moreover, a metagenomics analysis reveals that SC-HM ameliorates the dysbiosis of specific bacterial genera, including Bacteroides acidifaciens, Lactobacillus (L.) gasseri, Lactobacillus reuteri, and L. intestinalis, implying optimization of the microorganism's composition and abundance. These findings demonstrate that SC-HM is a potential IBD treatment candidate.
Collapse
Affiliation(s)
- Do-Wan Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Hye-Seon Jeong
- Division of Cosmetic Science and Technology, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Eunseo Kim
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Hyomin Lee
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, South Korea
| | - Chang-Hyung Choi
- Division of Cosmetic Science and Technology, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Sei-Jung Lee
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, South Korea.
| |
Collapse
|
45
|
Failla M, Lee J, Rasooly R, Apostolidis E. Evaluation of a Witch Hazel Extract for the Potential Prebiotic and Protective Effect on Select Lactiplantibacillus plantarum (Prev. Lactobacillus plantarum) Strains. Front Nutr 2022; 9:874666. [PMID: 35571953 PMCID: PMC9100581 DOI: 10.3389/fnut.2022.874666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Witch hazel extract has been evaluated in prior studies demonstrating the phenolic-mediated biofilm inhibition, toxin production inhibition, and growth inhibition in Staphylococcus aureus. In this study, we are evaluating the possible prebiotic and protective effect of witch hazel extract on select probiotic Lactiplantibacillus plantarum strains, namely L. plantarum LP 10241 and L. plantarum LPBAA-793. When the prebiotic effect was evaluated, we observed that the tested extract had prebiotic effect at the higher tested dose (0.5%) on LPBAA-793 strain (8.7 log CFU/mL after 18 h compared to 5.1 log CFU/mL with the control) and on LP 10241 strain (7.7 log CFU/mL after 18 h compared to 4.4 log CFU/mL with the control). For the evaluation of the protective effect of witch hazel extract on the select strains, we subjected nutrient depletion stress under aerobic conditions and monitored the cell death with and without addition of witch hazel extract. We observed that the tested extract had a significant protective effect on LPBAA-793 strain (4 log CFU/mL after 12 days, compared to no growth with control) and a slighter protective effect against LP 10241 strains (6.3 log CFU/mL in day 2 compared to 4.3 log CFU/mL with control). The results from this research provide for the first time the rationale that while witch hazel extract has significant antimicrobial, anti-toxin production and anti-biofilm activities on pathogenic microorganisms, it might play an important and positive role on health-beneficial probiotic bacteria.
Collapse
Affiliation(s)
- Morgan Failla
- Department of Chemistry and Food Science, Framingham State University, Framingham, MA, United States
| | - Jungyun Lee
- Department of Chemistry and Food Science, Framingham State University, Framingham, MA, United States
| | - Reuven Rasooly
- U.S. Department of Agriculture, Agricultural Research Service, Albany, CA, United States
- *Correspondence: Reuven Rasooly,
| | - Emmanouil Apostolidis
- Department of Chemistry and Food Science, Framingham State University, Framingham, MA, United States
- Emmanouil Apostolidis,
| |
Collapse
|
46
|
Phillippi DT, Daniel S, Nguyen KN, Penaredondo BA, Lund AK. Probiotics Function as Immunomodulators in the Intestine in C57Bl/6 Male Mice Exposed to Inhaled Diesel Exhaust Particles on a High-Fat Diet. Cells 2022; 11:cells11091445. [PMID: 35563751 PMCID: PMC9101602 DOI: 10.3390/cells11091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.
Collapse
Affiliation(s)
| | | | | | | | - Amie K. Lund
- Correspondence: ; Tel.: +1-(940)-369-8946; Fax: +1-(940)-565-4297
| |
Collapse
|
47
|
Li J, Chen D, Shen M. Tumor Microenvironment Shapes Colorectal Cancer Progression, Metastasis, and Treatment Responses. Front Med (Lausanne) 2022; 9:869010. [PMID: 35402443 PMCID: PMC8984105 DOI: 10.3389/fmed.2022.869010] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most devastating diseases that accounts for numerous deaths worldwide. Tumor cell-autonomous pathways, such as the oncogenic signaling activation, significantly contribute to CRC progression and metastasis. Recent accumulating evidence suggests that the CRC microenvironment also profoundly promotes or represses this process. As the roles of the tumor microenvironment (TME) in CRC progression and metastasis is gradually uncovered, the importance of these non-cell-autonomous signaling pathways is appreciated. However, we are still at the beginning of this TME function exploring process. In this review, we summarize the current understanding of the TME in CRC progression and metastasis by focusing on the gut microbiota and host cellular and non-cellular components. We also briefly discuss TME-remodeling therapies in CRC.
Collapse
Affiliation(s)
- Jun Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dawei Chen
- Wayne State University School of Medicine, Detroit, MI, United States
| | - Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
- *Correspondence: Minhong Shen,
| |
Collapse
|
48
|
Huang J, Zhang J, Wang X, Jin Z, Zhang P, Su H, Sun X. Effect of Probiotics on Respiratory Tract Allergic Disease and Gut Microbiota. Front Nutr 2022; 9:821900. [PMID: 35295917 PMCID: PMC8920559 DOI: 10.3389/fnut.2022.821900] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Allergy is a hypersensitivity reaction triggered by specific cell or antibody-mediated immune mechanisms. Allergies have increased in industrialized countries in recent decades. The rise in allergic respiratory diseases such as allergic rhinitis (AR) and allergic asthma (AA) is a potential threat to public health. Searches were conducted using PubMed, Google Scholar and Medline using the following key terms: allergic rhinitis OR asthma AND probiotics, allergic airway inflammation AND immune disorders, probiotics OR gut microbiota AND allergic disease, probiotics AND inflammatory. Studies from all years were included, specifically those published within the last 10 years. Some review articles and their reference lists were searched to identify related articles. The role of microbiota in respiratory allergic diseases has attracted more and more attention. Pieces of evidence suggested that the development of allergic diseases causes a possible imbalance in the composition of the gut microbiota. Compared to colonized mice, germ-free mice exhibit exaggerated allergic airway responses, suggesting that microbial host interactions play an important role in the development of allergic diseases. Probiotics modulate both the innate and adaptive inflammatory immune responses, often used as dietary supplements to provide health benefits in gastrointestinal disorders. Probiotics may serve as immunomodulators and activators of host defense pathways. Besides, oral probiotics can modulate the immune response in the respiratory system. Recently, studies in humans and animals have demonstrated the role of probiotic in RA and AA. To understand the characterization, microbiota, and the potential role of probiotics intervention of AA/AR, this review provides an overview of clinical features of AA and AR, probiotics for the prevention and treatment of AR, AA, changes in gut microbiota, and their mechanisms of action.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zenghui Jin
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geratology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
49
|
Kocot AM, Jarocka-Cyrta E, Drabińska N. Overview of the Importance of Biotics in Gut Barrier Integrity. Int J Mol Sci 2022; 23:ijms23052896. [PMID: 35270039 PMCID: PMC8911280 DOI: 10.3390/ijms23052896] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
Increased gut permeability is suggested to be involved in the pathogenesis of a growing number of disorders. The altered intestinal barrier and the subsequent translocation of bacteria or bacterial products into the internal milieu of the human body induce the inflammatory state. Gut microbiota maintains intestinal epithelium integrity. Since dysbiosis contributes to increased gut permeability, the interventions that change the gut microbiota and correct dysbiosis are suggested to also restore intestinal barrier function. In this review, the current knowledge on the role of biotics (probiotics, prebiotics, synbiotics and postbiotics) in maintaining the intestinal barrier function is summarized. The potential outcome of the results from in vitro and animal studies is presented, and the need for further well-designed randomized clinical trials is highlighted. Moreover, we indicate the need to understand the mechanisms by which biotics regulate the function of the intestinal barrier. This review is concluded with the future direction and requirement of studies involving biotics and gut barrier.
Collapse
Affiliation(s)
- Aleksandra Maria Kocot
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Tuwima 10, 10-748 Olsztyn, Poland;
| | - Elżbieta Jarocka-Cyrta
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine, Collegium Medicum University of Warmia and Mazury, Regional Specialized Children’s Hospital, Żołnierska St. 18A, 10-561 Olsztyn, Poland;
| | - Natalia Drabińska
- Department of Chemistry and Biodynamics of Food, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
- Correspondence:
| |
Collapse
|
50
|
Boscaini S, Leigh SJ, Lavelle A, García-Cabrerizo R, Lipuma T, Clarke G, Schellekens H, Cryan JF. Microbiota and body weight control: Weight watchers within? Mol Metab 2022; 57:101427. [PMID: 34973469 PMCID: PMC8829807 DOI: 10.1016/j.molmet.2021.101427] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite several decades of research, managing body weight remains an unsolved clinical problem. Health problems associated with dysregulated body weight, such as obesity and cachexia, exhibit several gut microbiota alterations. There is an increased interest in utilising the gut microbiota for body weight control, as it responds to intervention and plays an important role in energy extraction from food, as well as biotransformation of nutrients. SCOPE OF THE REVIEW This review provides an overview of the role of the gut microbiota in the physiological and metabolic alterations observed in two body weight dysregulation-related disorders, namely obesity and cachexia. Second, we assess the available evidence for different strategies, including caloric restriction, intermittent fasting, ketogenic diet, bariatric surgery, probiotics, prebiotics, synbiotics, high-fibre diet, and fermented foods - effects on body weight and gut microbiota composition. This approach was used to give insights into the possible link between body weight control and gut microbiota configuration. MAJOR CONCLUSIONS Despite extensive associations between body weight and gut microbiota composition, limited success could be achieved in the translation of microbiota-related interventions for body weight control in humans. Manipulation of the gut microbiota alone is insufficient to alter body weight and future research is needed with a combination of strategies to enhance the effects of lifestyle interventions.
Collapse
Affiliation(s)
- Serena Boscaini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Timothy Lipuma
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|